Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 136(8)2023 04 15.
Article in English | MEDLINE | ID: mdl-37013443

ABSTRACT

Calcineurin, or protein phosphatase 2B (PP2B), the Ca2+ and calmodulin-activated phosphatase and target of immunosuppressants, has many substrates and functions that remain uncharacterized. By combining rapid proximity-dependent labeling with cell cycle synchronization, we mapped the spatial distribution of calcineurin in different cell cycle stages. While calcineurin-proximal proteins did not vary significantly between interphase and mitosis, calcineurin consistently associated with multiple centrosomal and/or ciliary proteins. These include POC5, which binds centrins in a Ca2+-dependent manner and is a component of the luminal scaffold that stabilizes centrioles. We show that POC5 contains a calcineurin substrate motif (PxIxIT type) that mediates calcineurin binding in vivo and in vitro. Using indirect immunofluorescence and ultrastructure expansion microscopy, we demonstrate that calcineurin colocalizes with POC5 at the centriole, and further show that calcineurin inhibitors alter POC5 distribution within the centriole lumen. Our discovery that calcineurin directly associates with centriolar proteins highlights a role for Ca2+ and calcineurin signaling at these organelles. Calcineurin inhibition promotes elongation of primary cilia without affecting ciliogenesis. Thus, Ca2+ signaling within cilia includes previously unknown functions for calcineurin in maintenance of cilia length, a process that is frequently disrupted in ciliopathies.


Subject(s)
Calcineurin , Cilia , Calcineurin/metabolism , Cilia/metabolism , Calcium/metabolism , Centrosome/metabolism , Centrioles/metabolism , Proteins/metabolism
2.
Cell Syst ; 10(3): 254-264.e9, 2020 03 25.
Article in English | MEDLINE | ID: mdl-32191875

ABSTRACT

Half of the bacteria in the human gut microbiome are lysogens containing integrated prophages, which may activate in stressful immune environments. Although lysogens are likely to be phagocytosed by macrophages, whether prophage activation occurs or influences the outcome of bacterial infection remains unexplored. To study the dynamics of bacteria-phage interactions in living cells-in particular, the macrophage-triggered induction and lysis of dormant prophages in the phagosome-we adopted a tripartite system where murine macrophages engulf E. coli, which are lysogenic with an engineered bacteriophage λ, containing a fluorescent lysis reporter. Pre-induced prophages are capable of lysing the host bacterium and propagating infection to neighboring bacteria in the same phagosome. A non-canonical pathway, mediated by PhoP, is involved with the native λ phage induction inside phagocytosed E. coli. These findings suggest two possible mechanisms by which induced prophages may function to aid the bactericidal activity of macrophages.


Subject(s)
Lysogeny/physiology , Molecular Imaging/methods , Virus Activation/physiology , Animals , Bacteria , Bacteriophage lambda/physiology , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Gastrointestinal Microbiome , Genetic Engineering/methods , HEK293 Cells , Humans , Macrophages/metabolism , Mice , Prophages/metabolism , Prophages/physiology , RAW 264.7 Cells
3.
PLoS One ; 13(10): e0204957, 2018.
Article in English | MEDLINE | ID: mdl-30304739

ABSTRACT

Epithelial cell adhesion molecule (EpCAM) is a glycoprotein on the surface of epithelial cells that is essential for intestinal epithelial integrity and expressed at high levels in many epithelial derived cancers and circulating tumor cells. Here we show the effect of EpCAM levels on migration of Madin-Darby-Canine Kidney (MDCK) epithelial cells. MDCK cells depleted of EpCAM show increased activation of extracellular signal-regulated kinase (ERK) and of myosin, and increased cell spreading and epithelial sheet migration into a gap. In contrast, over-expression of EpCAM inhibits ERK and myosin activation, and slows epithelial sheet migration. Loss of EpCAM is rescued by EpCAM-YFP mutated in the extracellular domain required for cis-dimerization whereas EpCAM-YFP with a mutation that inhibits Claudin-7 interaction cannot rescue increased ERK, myosin activation, and increased migration in EpCAM-depleted cells. In summary, these results indicate that interaction of EpCAM and Claudin-7 at the cell surface negatively regulates epithelial migration by inhibiting ERK and actomyosin contractility.


Subject(s)
Cell Adhesion/physiology , Claudins/metabolism , Epithelial Cell Adhesion Molecule/metabolism , Actins/metabolism , Animals , Cell Movement/drug effects , Claudins/chemistry , Dimerization , Dogs , Epithelial Cell Adhesion Molecule/antagonists & inhibitors , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatocyte Growth Factor/pharmacology , Madin Darby Canine Kidney Cells/cytology , Madin Darby Canine Kidney Cells/metabolism , Microscopy, Confocal , Myosins/antagonists & inhibitors , Myosins/metabolism , Phosphorylation/drug effects , Protein Domains , RNA Interference , RNA, Small Interfering/metabolism , Time-Lapse Imaging
4.
Mol Biol Cell ; 25(7): 977-91, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24501426

ABSTRACT

ß-Catenin is a multifunctional protein with critical roles in cell-cell adhesion, Wnt signaling, and the centrosome cycle. Whereas the regulation of ß-catenin in cell-cell adhesion and Wnt signaling are well understood, how ß-catenin is regulated at the centrosome is not. NIMA-related protein kinase 2 (Nek2), which regulates centrosome disjunction/splitting, binds to and phosphorylates ß-catenin. Using in vitro and cell-based assays, we show that Nek2 phosphorylates the same regulatory sites in the N-terminus of ß-catenin as glycogen synthase kinase 3ß (GSK3ß), which are recognized by a specific phospho-S33/S37/T41 antibody, as well as additional sites. Nek2 binding to ß-catenin appears to inhibit binding of the E3 ligase ß-TrCP and prevents ß-catenin ubiquitination and degradation. Thus ß-catenin phosphorylated by Nek2 is stabilized and accumulates at centrosomes in mitosis. We further show that polo-like kinase 1 (Plk1) regulates Nek2 phosphorylation and stabilization of ß-catenin. Taken together, these results identify a novel mechanism for regulating ß-catenin stability that is independent of GSK3ß and provide new insight into a pathway involving Plk1, Nek2, and ß-catenin that regulates the centrosome cycle.


Subject(s)
Cell Cycle Proteins/metabolism , Centrosome/metabolism , Mitosis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , beta Catenin/metabolism , Casein Kinase I/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HCT116 Cells , HEK293 Cells , Humans , NIMA-Related Kinases , Phosphorylation , Protein Stability , Sequence Deletion , Serine/metabolism , Spindle Apparatus/metabolism , Polo-Like Kinase 1
5.
J Cell Sci ; 125(Pt 5): 1247-58, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22399805

ABSTRACT

Regulation of the microtubule- and actin-binding protein adenomatous polyposis coli (APC) is crucial for the formation of cell extensions in many cell types. This process requires inhibition of glycogen synthase kinase-3ß (GSK-3ß), which otherwise phosphorylates APC and decreases APC-mediated microtubule bundling. Although it is assumed, therefore, that APC phosphorylation is decreased during initiation of cell extensions, the phosphorylation state of APC has never been analyzed directly. We show here that NGF- and EGF-induced initial cell extensions result in APC phosphorylation by the MAPK/ERK pathway, which, in parallel with inhibition of GSK-3ß, promotes localization of APC to the tip of cell extensions. Whereas GSK-3ß inhibition promotes APC binding and stabilization of microtubules, we show that phosphorylation by ERK inhibits the interaction of APC with F-actin, and APC-mediated F-actin bundling, but not APC-mediated microtubule bundling, in vitro. These results identify a previously unknown APC regulatory pathway during growth-factor-induced cell extension, and indicate that the GSK-3ß and ERK pathways act in parallel to regulate interactions between APC and the cytoskeleton during the formation of cell extensions.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cell Surface Extensions/physiology , Cytoskeleton/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycogen Synthase Kinase 3/metabolism , Actins/metabolism , Animals , Cytoskeletal Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Microtubules/metabolism , Nerve Growth Factor/metabolism , PC12 Cells , Phosphorylation , RNA Interference , RNA, Small Interfering , Rats , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism
6.
J Cell Sci ; 123(Pt 18): 3125-35, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20736306

ABSTRACT

beta-Catenin has important roles in cell-cell adhesion and in the regulation of gene transcription. Mutations that stabilize beta-catenin are common in cancer, but it remains unclear how these mutations contribute to cancer progression. beta-Catenin is also a centrosomal component involved in centrosome separation. Centrosomes nucleate interphase microtubules and the bipolar mitotic spindle in normal cells, but their organization and function in human cancers are abnormal. Here, we show that expression of stabilized mutant beta-catenin, which mimics mutations found in cancer, results in extra non-microtubule nucleating structures that contain a subset of centrosome proteins including gamma-tubulin and centrin, but not polo-like kinase 4 (Plk4), SAS-6 or pericentrin. A transcriptionally inactive form of beta-catenin also gives rise to abnormal structures of centrosome proteins. HCT116 human colon cancer cell lines, from which the mutant beta-catenin allele has been deleted, have reduced numbers of cells with abnormal centrosome structures and S-phase-arrested, amplified centrosomes. RNAi-mediated depletion of beta-catenin from centrosomes inhibits S-phase-arrested amplification of centrosomes. These results indicate that beta-catenin is required for centrosome amplification, and mutations in beta-catenin might contribute to the formation of abnormal centrosomes observed in cancers.


Subject(s)
Centrosome/chemistry , Centrosome/metabolism , Neoplasms/metabolism , beta Catenin/metabolism , Animals , Cell Cycle , Cell Line, Tumor , Dogs , Humans , Neoplasms/genetics , Tubulin/genetics , Tubulin/metabolism , beta Catenin/genetics
7.
Adv Exp Med Biol ; 656: 65-74, 2009.
Article in English | MEDLINE | ID: mdl-19928353

ABSTRACT

Adenomatous polyposis coli (APC) is a multifunctional protein commonly mutated in colon cancer. APC contains binding sites for multiple proteins with diverse roles in signaling and the structural and functional organization of cells. Recent evidence suggests roles for APC and some of its binding partners in regulating microtubules in mitosis. APC localizes to three key locations in mitosis: kinetochores, the cortex and centrosomes. Here, we discuss possible mechanisms for APC function at these sites and suggest new pathways by which APC mutations promote tumorigenesis.


Subject(s)
Adenomatous Polyposis Coli Protein/physiology , Microtubules/metabolism , Mitosis/physiology , Adenomatous Polyposis Coli Protein/metabolism , Animals , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drosophila , Genes, APC , Humans , Kinetochores/metabolism , Mutation , Saccharomycetales
8.
Semin Cell Dev Biol ; 19(3): 245-51, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18387324

ABSTRACT

In response to extracellular signals during embryonic development, cells undergo directional movements to specific sites and establish proper connections to other cells to form organs and tissues. Cell extension and migration in the direction of extracellular cues is mediated by the actin and microtubule cytoskeletons, and recent results have shed new light on how these pathways are activated by neurotrophins, Wnt or extracellular matrix. These signals lead to modifications of microtubule-associated proteins (MAPs) and point to glycogen synthase kinase (GSK) 3beta as a key regulator of microtubule function during directional migration. This review will summarize these results and then focus on the role of microtubule-binding protein adenomatous polyposis coli (APC) in neuronal polarization and directed migration, and on its regulation by GSK3beta.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cell Movement , Cell Polarity , Microtubules/metabolism , Neurons/cytology , Adenomatous Polyposis Coli Protein/chemistry , Animals , Humans , Microtubule-Associated Proteins/metabolism
9.
Mol Cell Biol ; 28(7): 2283-94, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18212055

ABSTRACT

Centrosomes nucleate and organize interphase microtubules and are instrumental in mitotic bipolar spindle assembly, ensuring orderly cell cycle progression with accurate chromosome segregation. We report that the multifunctional structural protein 4.1R localizes at centrosomes to distal/subdistal regions of mature centrioles in a cell cycle-dependent pattern. Significantly, 4.1R-specific depletion mediated by RNA interference perturbs subdistal appendage proteins ninein and outer dense fiber 2/cenexin at mature centrosomes and concomitantly reduces interphase microtubule anchoring and organization. 4.1R depletion causes G(1) accumulation in p53-proficient cells, similar to depletion of many other proteins that compromise centrosome integrity. In p53-deficient cells, 4.1R depletion delays S phase, but aberrant ninein distribution is not dependent on the S-phase delay. In 4.1R-depleted mitotic cells, efficient centrosome separation is reduced, resulting in monopolar spindle formation. Multipolar spindles and bipolar spindles with misaligned chromatin are also induced by 4.1R depletion. Notably, all types of defective spindles have mislocalized NuMA (nuclear mitotic apparatus protein), a 4.1R binding partner essential for spindle pole focusing. These disruptions contribute to lagging chromosomes and aberrant microtubule bridges during anaphase/telophase. Our data provide functional evidence that 4.1R makes crucial contributions to the structural integrity of centrosomes and mitotic spindles which normally enable mitosis and anaphase to proceed with the coordinated precision required to avoid pathological events.


Subject(s)
Cell Cycle/physiology , Centrioles/chemistry , Centrosome/ultrastructure , Cytoskeletal Proteins/physiology , Membrane Proteins/physiology , Spindle Apparatus/ultrastructure , Anaphase/physiology , Antigens, Nuclear/analysis , Cell Cycle Proteins , Cell Line/ultrastructure , Centrosome/metabolism , Chromosome Segregation/physiology , Cytoskeletal Proteins/analysis , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Down-Regulation , HeLa Cells/ultrastructure , Humans , Interphase/physiology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Microtubules/ultrastructure , Nuclear Matrix-Associated Proteins/analysis , Nuclear Proteins/analysis , RNA Interference , RNA, Small Interfering/pharmacology , Spindle Apparatus/chemistry , Spindle Apparatus/metabolism , Tumor Suppressor Protein p53/physiology
10.
Genes Dev ; 22(1): 91-105, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18086858

ABSTRACT

beta-Catenin plays important roles in cell adhesion and gene transcription, and has been shown recently to be essential for the establishment of a bipolar mitotic spindle. Here we show that beta-catenin is a component of interphase centrosomes and that stabilization of beta-catenin, mimicking mutations found in cancers, induces centrosome splitting. Centrosomes are held together by a dynamic linker regulated by Nek2 kinase and its substrates C-Nap1 (centrosomal Nek2-associated protein 1) and Rootletin. We show that beta-catenin binds to and is phosphorylated by Nek2, and is in a complex with Rootletin. In interphase, beta-catenin colocalizes with Rootletin between C-Nap1 puncta at the proximal end of centrioles, and this localization is dependent on C-Nap1 and Rootletin. In mitosis, when Nek2 activity increases, beta-catenin localizes to centrosomes at spindle poles independent of Rootletin. Increased Nek2 activity disrupts the interaction of Rootletin with centrosomes and results in binding of beta-catenin to Rootletin-independent sites on centrosomes, an event that is required for centrosome separation. These results identify beta-catenin as a component of the intercentrosomal linker and define a new function for beta-catenin as a key regulator of mitotic centrosome separation.


Subject(s)
Centrosome/metabolism , Protein Serine-Threonine Kinases/metabolism , beta Catenin/metabolism , Animals , Armadillo Domain Proteins/metabolism , Cells, Cultured , Centrosome/enzymology , Cytoskeletal Proteins/analysis , Cytoskeletal Proteins/metabolism , Dogs , Humans , Interphase , NIMA-Related Kinases , Proteins/analysis , Proteins/metabolism , beta Catenin/analysis , tRNA Methyltransferases
11.
J Cell Sci ; 118(Pt 24): 5699-708, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16303851

ABSTRACT

Neuronal morphogenesis involves the initial formation of neurites and then differentiation of neurites into axons and dendrites. The mechanisms underlying neurite formation are poorly understood. A candidate protein for controlling neurite extension is the adenomatous polyposis coli (APC) protein, which regulates membrane extensions, microtubules and beta-catenin-mediated transcription downstream of Wnt signaling. APC is enriched at the tip of several neurites of unpolarized hippocampal neurons and the tip of only the long axon in polarized hippocampal neurons. Significantly, APC localized to the tip of only one neurite, marked by dephospho-tau as the future axon, before that neurite had grown considerably longer than other neurites. To determine whether neurite outgrowth was affected by beta-catenin accumulation and signaling, a stabilized beta-catenin mutant was expressed in PC12 cells, and neurite formation was measured. Stabilized beta-catenin mutants accumulated in APC clusters and inhibited neurite formation and growth. Importantly, these effects were also observed was independently of the gene transcriptional activity of beta-catenin. These results indicate that APC is involved in both early neurite outgrowth and increased growth of the future axon, and that beta-catenin has a structural role in inhibiting APC function in neurite growth.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Hippocampus/metabolism , Neurites/metabolism , Signal Transduction/physiology , beta Catenin/metabolism , Animals , Gene Expression , Mutation , PC12 Cells , Rats , beta Catenin/genetics
12.
J Cell Sci ; 117(Pt 7): 1117-28, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14970257

ABSTRACT

Adenomatous polyposis coli (APC) and End-binding protein 1 (EB1) localize to centrosomes independently of cytoplasmic microtubules (MTs) and purify with centrosomes from mammalian cell lines. Localization of EB1 to centrosomes is independent of its MT binding domain and is mediated by its C-terminus. Both APC and EB1 preferentially localize to the mother centriole and EB1 forms a cap at the end of the mother centriole that contains the subdistal appendages as defined by epsilon-tubulin localization. Like endogenous APC and EB1, fluorescent protein fusions of APC and EB1 localize preferentially to the mother centriole. Depletion of EB1 by RNA interference reduces MT minus-end anchoring at centrosomes and delays MT regrowth from centrosomes. In summary, our data indicate that APC and EB1 are functional components of mammalian centrosomes and that EB1 is important for anchoring cytoplasmic MT minus ends to the subdistal appendages of the mother centriole.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Centrioles/metabolism , Centrosome/metabolism , Microtubule-Associated Proteins/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , COS Cells , Cell Line , Dogs , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Microtubule-Associated Proteins/genetics , Microtubules/drug effects , Microtubules/metabolism , Nocodazole/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
Bioessays ; 24(9): 771-4, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12210511

ABSTRACT

Somatic or inherited mutations in the adenomatous polyposis coli (APC) gene are a frequent cause of colorectal cancer in humans. APC protein has an important tumor suppression function to reduce cellular levels of the signaling protein beta-catenin and, thereby, inhibit beta-catenin and T-cell-factor-mediated gene expression. In addition, APC protein binds to microtubules in vertebrate cells and localizes to actin-rich adherens junctions in epithelial cells of the fruit fly Drosophila (Fig. 1). Very little is known, however, about the function of these cytoskeletal associations. Recently, Hamada and Bienz have described a potential role for Drosophila E-APC in cellular adhesion, which offers new clues to APC function in embryonic development, and potentially colorectal adenoma formation and tumor progression in humans.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Mutation , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Cell Adhesion , Colorectal Neoplasms/metabolism , Drosophila , Humans , Models, Biological
14.
J Cell Sci ; 115(Pt 8): 1583-90, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11950877

ABSTRACT

End-binding protein (EB) 1 binds to the C-terminus of adenomatous polyposis coli (APC) protein and to the plus ends of microtubules (MT) and has been implicated in the regulation of APC accumulation in cortical clusters at the tip of extending membranes. We investigated which APC domains are involved in cluster localization and whether binding to EB1 or MTs is essential for APC cluster localization. Armadillo repeats of APC that lack EB1- and MT-binding domains are necessary and sufficient for APC localization in cortical clusters; an APC fragment lacking the armadillo repeats, but containing MT- and EB1-binding domains, does not localize to the cortical clusters but instead co-aligns with MTs throughout the cell. Significantly, analysis of endogenous proteins reveals that EB1 does not accumulate in the APC clusters. However, overexpressed EB1 does accumulate in APC clusters; the APC-binding domain in EB1 is located in the C-terminal region of EB1 between amino acids 134 and 268. Overexpressed APC- or MT-binding domains of EB1 localize to APC cortical clusters and MT, respectively, without affecting APC cluster formation itself. These results show that localization of APC in cortical clusters is different from that of EB1 at MT plus ends and appears to be independent of EB1.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cell Membrane/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Adenomatous Polyposis Coli Protein/chemistry , Adenomatous Polyposis Coli Protein/genetics , Animals , Cell Line , Dogs , Genes, Reporter , Microscopy, Fluorescence , Microtubule-Associated Proteins/genetics , Microtubules/chemistry , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...